Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
1.
Comp Immunol Microbiol Infect Dis ; 109: 102183, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38640700

ABSTRACT

Henipavirus (HNV) is well known for two zoonotic viruses in the genus, Hendra virus (HeV) and Nipah virus (NiV), which pose serious threat to human and animal health. In August 2022, a third zoonotic virus in the genus Henipavirus, Langya virus (LayV), was discovered in China. The emergence of HeV, NiV, and LayV highlights the persistent threat of HNV to human and animal health. In addition to the above three HNVs, new species within this genus are still being discovered. Although they have not yet caused a pandemic in humans or livestock, they still have the risk of spillover as a potential threat to the health of humans and animals. It's important to understand the infection and transmission of different HNV in animals for the prevention and control of current or future HNV epidemics. Therefore, this review mainly summarizes the animal origin, animal infection and transmission of HNV that have been found worldwide, and further analyzes and summarizes the rules of infection and transmission, so as to provide a reference for relevant scientific researchers. Furthermore, it can provide a direction for epidemic prevention and control, and animal surveillance to reduce the risk of the global pandemic of HNV.


Subject(s)
Henipavirus Infections , Henipavirus , Animals , Henipavirus Infections/transmission , Henipavirus Infections/epidemiology , Henipavirus Infections/veterinary , Henipavirus Infections/virology , Humans , Zoonoses/transmission , Zoonoses/virology , Viral Zoonoses/transmission , Viral Zoonoses/virology , Nipah Virus/pathogenicity , Hendra Virus
3.
PLoS One ; 17(2): e0263834, 2022.
Article in English | MEDLINE | ID: mdl-35143571

ABSTRACT

Disease associated with Nipah virus infection causes a devastating and often fatal spectrum of syndromes predominated by both respiratory and neurologic conditions. Additionally, neurologic sequelae may manifest months to years later after virus exposure or apparent recovery. In the two decades since this disease emerged, much work has been completed in an attempt to understand the pathogenesis and facilitate development of medical countermeasures. Here we provide detailed organ system-specific pathologic findings following exposure of four African green monkeys to 2.41×105 pfu of the Malaysian strain of Nipah virus. Our results further substantiate the African green monkey as a model of human Nipah virus disease, by demonstrating both the respiratory and neurologic components of disease. Additionally, we demonstrate that a chronic phase of disease exists in this model, that may provide an important opportunity to study the enigmatic late onset and relapse encephalitis as it is described in human disease.


Subject(s)
Encephalitis, Viral/pathology , Henipavirus Infections/pathology , Lung Diseases/virology , Nipah Virus/pathogenicity , Animals , Chlorocebus aethiops , Disease Models, Animal , Lung Diseases/pathology , Malaysia , Male , Nipah Virus/classification
4.
Lancet Infect Dis ; 22(1): e13-e27, 2022 01.
Article in English | MEDLINE | ID: mdl-34735799

ABSTRACT

Henipaviruses, including Nipah virus, are regarded as pathogens of notable epidemic potential because of their high pathogenicity and the paucity of specific medical countermeasures to control infections in humans. We review the evidence of medical countermeasures against henipaviruses and project their cost in a post-COVID-19 era. Given the sporadic and unpredictable nature of henipavirus outbreaks, innovative strategies will be needed to circumvent the infeasibility of traditional phase 3 clinical trial regulatory pathways. Stronger partnerships with scientific institutions and regulatory authorities in low-income and middle-income countries can inform coordination of appropriate investments and development of strategies and normative guidelines for the deployment and equitable use of multiple medical countermeasures. Accessible measures should include global, regional, and endemic in-country stockpiles of reasonably priced small molecules, monoclonal antibodies, and vaccines as part of a combined collection of products that could help to control henipavirus outbreaks and prevent future pandemics.


Subject(s)
Disease Outbreaks/prevention & control , Henipavirus Infections/drug therapy , Henipavirus/pathogenicity , Medical Countermeasures , Public Health , Animals , COVID-19/prevention & control , Chiroptera/virology , Clinical Trials, Phase III as Topic , Henipavirus/classification , Henipavirus Infections/prevention & control , Henipavirus Infections/transmission , Humans , Nipah Virus/pathogenicity , SARS-CoV-2/pathogenicity
6.
Viruses ; 13(7)2021 07 02.
Article in English | MEDLINE | ID: mdl-34372504

ABSTRACT

Pandemics are a consequence of a series of processes that span scales from viral biology at 10-9 m to global transmission at 106 m. The pathogen passes from one host species to another through a sequence of events that starts with an infected reservoir host and entails interspecific contact, innate immune responses, receptor protein structure within the potential host, and the global spread of the novel pathogen through the naive host population. Each event presents a potential barrier to the onward passage of the virus and should be characterized with an integrated transdisciplinary approach. Epidemic control is based on the prevention of exposure, infection, and disease. However, the ultimate pandemic prevention is prevention of the spillover event itself. Here, we focus on the potential for preventing the spillover of henipaviruses, a group of viruses derived from bats that frequently cross species barriers, incur high human mortality, and are transmitted among humans via stuttering chains. We outline the transdisciplinary approach needed to prevent the spillover process and, therefore, future pandemics.


Subject(s)
Chiroptera/virology , Global Health , Henipavirus Infections/prevention & control , Henipavirus/pathogenicity , Pandemics/prevention & control , Virus Diseases/prevention & control , Zoonoses/virology , Animals , Henipavirus Infections/epidemiology , Henipavirus Infections/immunology , Henipavirus Infections/transmission , Host Specificity , Humans , Immunity, Innate , Nipah Virus/pathogenicity , Virus Diseases/immunology , Virus Diseases/transmission , Zoonoses/prevention & control , Zoonoses/transmission
7.
J Gen Virol ; 102(1)2021 01.
Article in English | MEDLINE | ID: mdl-33054904

ABSTRACT

Although enveloped viruses canonically mediate particle entry through virus-cell fusion, certain viruses can spread by cell-cell fusion, brought about by receptor engagement and triggering of membrane-bound, viral-encoded fusion proteins on the surface of cells. The formation of pathogenic syncytia or multinucleated cells is seen in vivo, but their contribution to viral pathogenesis is poorly understood. For the negative-strand paramyxoviruses respiratory syncytial virus (RSV) and Nipah virus (NiV), cell-cell spread is highly efficient because their oligomeric fusion protein complexes are active at neutral pH. The recently emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has also been reported to induce syncytia formation in infected cells, with the spike protein initiating cell-cell fusion. Whilst it is well established that fusion protein-specific antibodies can block particle attachment and/or entry into the cell (canonical virus neutralization), their capacity to inhibit cell-cell fusion and the consequences of this neutralization for the control of infection are not well characterized, in part because of the lack of specific tools to assay and quantify this activity. Using an adapted bimolecular fluorescence complementation assay, based on a split GFP-Renilla luciferase reporter, we have established a micro-fusion inhibition test (mFIT) that allows the identification and quantification of these neutralizing antibodies. This assay has been optimized for high-throughput use and its applicability has been demonstrated by screening monoclonal antibody (mAb)-mediated inhibition of RSV and NiV fusion and, separately, the development of fusion-inhibitory antibodies following NiV vaccine immunization in pigs. In light of the recent emergence of coronavirus disease 2019 (COVID-19), a similar assay was developed for SARS-CoV-2 and used to screen mAbs and convalescent patient plasma for fusion-inhibitory antibodies. Using mFITs to assess antibody responses following natural infection or vaccination is favourable, as this assay can be performed entirely at low biocontainment, without the need for live virus. In addition, the repertoire of antibodies that inhibit cell-cell fusion may be different to those that inhibit particle entry, shedding light on the mechanisms underpinning antibody-mediated neutralization of viral spread.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antibodies, Viral/pharmacology , COVID-19/diagnosis , Henipavirus Infections/diagnosis , High-Throughput Screening Assays , Respiratory Syncytial Virus Infections/diagnosis , Viral Fusion Proteins/antagonists & inhibitors , Animals , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/metabolism , Antibodies, Viral/isolation & purification , Antibodies, Viral/metabolism , COVID-19/immunology , COVID-19/virology , Cell Fusion , Convalescence , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Henipavirus Infections/immunology , Henipavirus Infections/virology , Humans , Immune Sera/chemistry , Luciferases/genetics , Luciferases/metabolism , Models, Molecular , Nipah Virus/immunology , Nipah Virus/pathogenicity , Protein Conformation , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/pathogenicity , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Swine , Viral Fusion Protein Inhibitors/chemistry , Viral Fusion Protein Inhibitors/metabolism , Viral Fusion Protein Inhibitors/pharmacology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology
8.
Infect Genet Evol ; 85: 104442, 2020 11.
Article in English | MEDLINE | ID: mdl-32622923

ABSTRACT

Little is known about the genetic features of Nipah virus (NiV) associated with virulence and transmission. Herein, phylogenetic and genetic analyses for all available NiV strains revealed sequence variations between the two genetic lineages of NiV with pathogenic differences, as well as among different strains within Bangladesh lineage. A total of 143 conserved amino acid differences, distributed among viral nucleocapsid (N), phosphoprotein (P), matrix protein (M), fusion protein (F) and glycoprotein (G), were revealed. Structural modeling revealed one key substitution (S3554N) in the viral G protein that might mediate a 12-amino-acid structural change from a loop into a ß sheet. Multiple key amino acids substitutions in viral G protein were observed, which may alter viral fitness and transmissibility from bats to humans.


Subject(s)
Genetic Variation , Henipavirus Infections/transmission , Henipavirus Infections/virology , Nipah Virus/genetics , Nipah Virus/pathogenicity , Phylogeny , Viral Proteins/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Bangladesh , Chiroptera/virology , Evolution, Molecular , Humans , Malaysia , Virulence
10.
mSphere ; 5(4)2020 07 08.
Article in English | MEDLINE | ID: mdl-32641430

ABSTRACT

Nipah disease is listed as one of the WHO priority diseases that pose the greatest public health risk due to their epidemic potential. More than 200 experts from around the world convened in Singapore last year to mark the 20th anniversary of the first Nipah virus outbreaks in Malaysia and Singapore. Most of these experts are now involved in responding to the coronavirus disease 2019 (COVID-19) pandemic. Here, members of the Organizing Committee of the 2019 Nipah Virus International Conference review highlights from the Nipah@20 Conference and reflect on key lessons learned from Nipah that could be applied to the understanding of the COVID-19 pandemic and to preparedness against future emerging infectious diseases (EIDs) of pandemic potential.


Subject(s)
Henipavirus Infections , Nipah Virus/pathogenicity , Animals , Betacoronavirus/pathogenicity , COVID-19 , Congresses as Topic , Coronavirus Infections/diagnosis , Coronavirus Infections/prevention & control , Coronavirus Infections/therapy , Henipavirus Infections/diagnosis , Henipavirus Infections/prevention & control , Henipavirus Infections/therapy , Humans , Pandemics/prevention & control , Pneumonia, Viral/diagnosis , Pneumonia, Viral/prevention & control , Pneumonia, Viral/therapy , SARS-CoV-2 , Zoonoses/epidemiology
11.
Microb Pathog ; 141: 103976, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31940461

ABSTRACT

The Nipah Virus (NiV) was first isolated during a 1998-9 outbreak in Malaysia. The outbreak initially infected farm pigs and then moved to humans from pigs with a case-fatality rate (CFR) of about 40%. After 2001, regular outbreaks occurred with higher CFRs (~71%, 2001-5, ~93%, 2008-12). The spread arose from drinking virus-laden palm date sap and human-to-human transmission. Intrinsic disorder analysis revealed strong correlation between the percentage of disorder in the N protein and CFR (Regression: r2 = 0.93, p < 0.01, ANOVA: p < 0.01). Distinct disorder and, therefore, genetic differences can be found in all three group of strains. The fact that the transmission modes of the Malaysia strain are different from those of the Bangladesh strains suggests that the correlations may also be linked to the modes of viral transmission. Analysis of the NiV and related viruses suggests links between modes of transmission and disorder of not just the N protein but, also, of M shell protein. The links among shell disorder, transmission modes, and virulence suggest mechanisms by which viruses are attenuated as they passed through different cell hosts from different animal species. These have implications for development of vaccines and epidemiological molecular analytical tools to contain outbreaks.


Subject(s)
Henipavirus Infections/virology , Nipah Virus/pathogenicity , Amino Acid Sequence , Animals , Disease Outbreaks , Disease Susceptibility , Evolution, Molecular , Genome, Viral , Henipavirus Infections/epidemiology , Humans , Models, Biological , Mortality , Nipah Virus/classification , Nipah Virus/genetics , Phylogeny , Protein Conformation , Sequence Analysis, DNA , Swine , Swine Diseases/epidemiology , Swine Diseases/virology , Viral Proteins/chemistry , Viral Proteins/genetics , Virulence
12.
Transbound Emerg Dis ; 67(1): 121-132, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31408582

ABSTRACT

Since its first emergence in 1998 in Malaysia, Nipah virus (NiV) has become a great threat to domestic animals and humans. Sporadic outbreaks associated with human-to-human transmission caused hundreds of human fatalities. Here, we collected all available NiV sequences and combined phylogenetics, molecular selection, structural biology and receptor analysis to study the emergence and adaptive evolution of NiV. NiV can be divided into two main lineages including the Bangladesh and Malaysia lineages. We formly confirmed a significant association with geography which is probably the result of long-term evolution of NiV in local bat population. The two NiV lineages differ in many amino acids; one change in the fusion protein might be involved in its activation via binding to the G protein. We also identified adaptive and positively selected sites in many viral proteins. In the receptor-binding G protein, we found that sites 384, 386 and especially 498 of G protein might modulate receptor-binding affinity and thus contribute to the host jump from bats to humans via the adaption to bind the human ephrin-B2 receptor. We also found that site 1645 in the connector domain of L was positive selected and involved in adaptive evolution; this site might add methyl groups to the cap structure present at the 5'-end of the RNA and thus modulate its activity. This study provides insight to assist the design of early detection methods for NiV to assess its epidemic potential in humans.


Subject(s)
Adaptation, Biological , Chiroptera/virology , Disease Outbreaks , Henipavirus Infections/virology , Nipah Virus/genetics , Polymorphism, Genetic , Animals , Bangladesh/epidemiology , Biological Evolution , Computational Biology , Geography , Henipavirus Infections/epidemiology , Henipavirus Infections/transmission , Host Specificity , Humans , Malaysia/epidemiology , Models, Molecular , Nipah Virus/isolation & purification , Nipah Virus/pathogenicity , Nipah Virus/physiology , Phylogeny , Viral Proteins/genetics
13.
Emerg Infect Dis ; 26(1): 104-113, 2020 01.
Article in English | MEDLINE | ID: mdl-31855143

ABSTRACT

We conducted an in-depth characterization of the Nipah virus (NiV) isolate previously obtained from a Pteropus lylei bat in Cambodia in 2003 (CSUR381). We performed full-genome sequencing and phylogenetic analyses and confirmed CSUR381 is part of the NiV-Malaysia genotype. In vitro studies revealed similar cell permissiveness and replication of CSUR381 (compared with 2 other NiV isolates) in both bat and human cell lines. Sequence alignments indicated conservation of the ephrin-B2 and ephrin-B3 receptor binding sites, the glycosylation site on the G attachment protein, as well as the editing site in phosphoprotein, suggesting production of nonstructural proteins V and W, known to counteract the host innate immunity. In the hamster animal model, CSUR381 induced lethal infections. Altogether, these data suggest that the Cambodia bat-derived NiV isolate has high pathogenic potential and, thus, provide insight for further studies and better risk assessment for future NiV outbreaks in Southeast Asia.


Subject(s)
Chiroptera/virology , Henipavirus Infections/veterinary , Nipah Virus/pathogenicity , Animals , Cambodia , Genome, Viral/genetics , Henipavirus Infections/epidemiology , Henipavirus Infections/virology , Humans , Nipah Virus/genetics , Phylogeny , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Whole Genome Sequencing
14.
F1000Res ; 82019.
Article in English | MEDLINE | ID: mdl-31656582

ABSTRACT

Nipah virus (NiV) is a highly lethal zoonotic paramyxovirus that emerged at the end of last century as a human pathogen capable of causing severe acute respiratory infection and encephalitis. Although NiV provokes serious diseases in numerous mammalian species, the infection seems to be asymptomatic in NiV natural hosts, the fruit bats, which provide a continuous virus source for further outbreaks. Consecutive human-to-human transmission has been frequently observed during outbreaks in Bangladesh and India. NiV was shown to interfere with the innate immune response and interferon type I signaling, restraining the anti-viral response and permitting viral spread. Studies of adaptive immunity in infected patients and animal models have suggested an unbalanced immune response during NiV infection. Here, we summarize some of the recent studies of NiV pathogenesis and NiV-induced modulation of both innate and adaptive immune responses, as well as the development of novel prophylactic and therapeutic approaches, necessary to control this highly lethal emerging infection.


Subject(s)
Adaptive Immunity , Henipavirus Infections/immunology , Immunity, Innate , Nipah Virus/pathogenicity , Animals , Bangladesh , Humans , India , Nipah Virus/immunology
15.
Curr Pharm Des ; 25(12): 1392-1401, 2019.
Article in English | MEDLINE | ID: mdl-31258065

ABSTRACT

BACKGROUND: The recent Nipah virus (NiV) outbreak in India has caused a state of chaos, with potential to become the next international pandemic. There is still a great deal to learn about NiV for the development of a potent treatment against it. The NiV non-structural proteins play important roles in the lifecycle of the virus, with the RNA-dependent RNA-polymerase (RdRp) being a vital component in viral replication. In this study, we not only provide a comprehensive overview of all the literature concerning NiV, we also propose a model of the NiV RdRp and screen for potential inhibitors of the viral enzyme. METHODS: In this study, computational tools were utilized in the design of a NiV RdRp homology model. The active site of RdRp was then identified and potential inhibitors of the protein were discovered with the use of pharmacophore-based screening. RESULTS: Ramachandran plot analysis revealed a favourable model. Upon binding of nucleoside analog, 4'- Azidocytidine, active site residues Trp1714 and Ser1713 took part in stabilizing hydrogen bonds, while Thr1716, Ser1478, Ser1476 and Glu1465 contributed to hydrophobic interactions. Pharmacophore based screening yielded 18 hits, of which ZINC00085930 demonstrated the most optimal binding energy (-8.1 kcal/mol), validating its use for further analysis as an inhibitor of NiV. CONCLUSION: In this study we provide a critical guide, elucidating on the in silico requirements of the drug design and discovery process against NiV. This material lays a foundation for future research into the design and development of drugs that inhibit NiV.


Subject(s)
Antiviral Agents/pharmacology , Nipah Virus/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Catalytic Domain , Drug Design , Nipah Virus/pathogenicity , Nipah Virus/physiology , Virus Replication
16.
JCI Insight ; 4(14)2019 07 25.
Article in English | MEDLINE | ID: mdl-31341108

ABSTRACT

Nipah virus (NiV) is an emerging zoonotic paramyxovirus that causes highly lethal henipavirus encephalitis in humans. Survivors develop various neurologic sequelae, including late-onset and relapsing encephalitis, several months up to several years following initial infection. However, the underlying pathology and disease mechanisms of persistent neurologic complications remain unknown. Here, we demonstrate persistent NiV infection in the brains of grivets that survived experimental exposure to NiV. Encephalitis affected the entire brains, with the majority of NiV detected in the neurons and microglia of the brainstems, cerebral cortices, and cerebella. We identified the vascular endothelium in the brain as an initial target of NiV infection during the acute phase of disease, indicating a primary path of entry for NiV into the brain. Notably, we were unable to detect NiV anywhere else except the brains in the examined survivors. Our findings indicate that late-onset and relapsing encephalitis of NiV in human survivors may be due to viral persistence in the brain and shed light on the pathogenesis of chronic henipavirus encephalitis.


Subject(s)
Brain/virology , Communicable Diseases, Emerging/pathology , Henipavirus Infections/pathology , Nipah Virus/isolation & purification , Zoonoses/pathology , Animals , Brain/blood supply , Brain/pathology , Chlorocebus aethiops , Chronic Disease , Communicable Diseases, Emerging/mortality , Communicable Diseases, Emerging/virology , Disease Models, Animal , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , Henipavirus Infections/mortality , Henipavirus Infections/virology , Humans , Male , Nipah Virus/pathogenicity , Recurrence , Survivors , Zoonoses/mortality , Zoonoses/virology
17.
J Clin Virol ; 117: 19-26, 2019 08.
Article in English | MEDLINE | ID: mdl-31132674

ABSTRACT

BACKGROUND: A wide range of Nipah virus (NiV) encephalitis case fatality rates (CFR) have been reported. Data on the involvement of several potential risk factors in Nipah virus transmission remain controversial. We performed a systematic review and meta-analysis to estimate the pooled CFR of NiV encephalitis and to assess the risk factors for NiV infection. METHODS: Articles published up to the 27thof November 2018 in MedLine, Embase and Web of knowledge databases were considered for this study. We included cross-sectional, cohort, and case-control studies that have reported NiV CFR and/or risk factors. Data were pooled with random-effects model. This review was registered in the PROSPERO, CRD42018116242. FINDINGS: This global review included 22 citations (25 studies) including 2156, 1682, and 474 suspected, probable, and confirmed cases of NiV encephalitis, respectively. We determined a pooled CFR for NiV encephalitis at 61.0% (95% CI, 45.7-75.4; I² = 96.8%). Climbing trees (OR = 1.4; 95% CI; 1.0-1.9), male gender (OR = 1.5; 95% CI; 1.1-2.0), travel outside their own sub-district (OR = 2.0; 95% CI; 1.4-2.9), and exposure to date palm sap (DPS) (OR = 5.7; 95% CI; 3.8-8.6) or pigs (OR = 7.6; 95% CI; 1.2-45.4) were significantly associated with NiV infection. CONCLUSION: Findings from this study suggest that NiV Encephalitis is associated with a high CFR and that male gender, travel outside their sub-district, climbing trees, and exposure to pigs and DPS are associated with an increased risk of NiV encephalitis.


Subject(s)
Encephalitis, Viral/mortality , Henipavirus Infections/mortality , Nipah Virus/pathogenicity , Animals , Female , Humans , Male , Mortality , Risk Factors , Sex Characteristics , Zoonoses/epidemiology , Zoonoses/mortality
18.
PLoS Pathog ; 15(4): e1007733, 2019 04.
Article in English | MEDLINE | ID: mdl-31034506

ABSTRACT

Formation of cytoplasmic inclusion bodies (IBs) is a hallmark of infections with non-segmented negative-strand RNA viruses (order Mononegavirales). We show here that Nipah virus (NiV), a bat-derived highly pathogenic member of the Paramyxoviridae family, differs from mononegaviruses of the Rhabdo-, Filo- and Pneumoviridae families by forming two types of IBs with distinct localizations, formation kinetics, and protein compositions. IBs in the perinuclear region form rapidly upon expression of the nucleocapsid proteins. These IBperi are highly mobile and associate with the aggresome marker y-tubulin. IBperi can recruit unrelated overexpressed cytosolic proteins but do not contain the viral matrix (M) protein. Additionally, NiV forms an as yet undescribed IB population at the plasma membrane (IBPM) that is y-tubulin-negative but contains the M protein. Infection studies with recombinant NiV revealed that IBPM require the M protein for their formation, and most likely represent sites of NiV assembly and budding. The identification of this novel type of plasma membrane-associated IBs not only provides new insights into NiV biology and may open new avenues to develop novel antiviral approaches to treat these highly pathogenic viruses, it also provides a basis for a more detailed characterization of IBs and their role in virus assembly and replication in infections with other Mononegavirales.


Subject(s)
Cell Membrane/virology , Henipavirus Infections/virology , Inclusion Bodies, Viral/virology , Nipah Virus/pathogenicity , Viral Matrix Proteins/metabolism , Animals , Chlorocebus aethiops , Glycoproteins/metabolism , Henipavirus Infections/metabolism , Henipavirus Infections/pathology , Humans , Inclusion Bodies, Viral/metabolism , Inclusion Bodies, Viral/pathology , Vero Cells , Virus Assembly , Virus Internalization
19.
Sci Rep ; 9(1): 5230, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30914663

ABSTRACT

In 1998 an outbreak of fatal encephalitis among pig farm workers in Malaysia and Singapore led to the discovery of Nipah henipavirus (NiV), a novel paramyxovirus closely related to Hendra henipavirus with case fatality rates of nearly 40%. Following its initial emergence nearly annual outbreaks of NiV have occurred in Bangladesh with a different, NiV Bangladesh, genotype, where the role of pigs in its transmission remains unknown. The present study provides the first report on susceptibility of domestic pigs to NiV Bangladesh following experimental infection, characterizing acute and long-term phases of disease and pathogenesis. All pigs were successfully infected with NiV Bangladesh following oronasal inoculation, with viral shedding confirmed by a novel genotype-specific qRT-PCR in oral, nasal and rectal excretions and dissemination from the upper respiratory tract to the brain, lungs, and associated lymphatic tissues. Unlike previous NiV Malaysia findings in pigs, clinical signs were absent, viremia was undetectable throughout the study, and only low level neutralizing antibody titers were measured by 28/29 days post-NiV-B infection. Results obtained highlight the need for continued and enhanced NiV surveillance in pigs in endemic and at-risk regions, and raise questions regarding applicability of current serological assays to detect animals with previous NiV-B exposure.


Subject(s)
Henipavirus Infections , Nipah Virus/pathogenicity , Swine Diseases , Swine , Animals , Bangladesh/epidemiology , Henipavirus Infections/epidemiology , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Swine/metabolism , Swine/virology , Swine Diseases/epidemiology , Swine Diseases/metabolism , Swine Diseases/virology
20.
Viruses ; 11(3)2019 03 22.
Article in English | MEDLINE | ID: mdl-30909389

ABSTRACT

Syrian hamsters (Mesocricetus auratus) are a pathogenesis model for the Nipah virus (NiV), and we sought to determine if they are also susceptible to the Cedar virus (CedPV). Following intranasal inoculation with CedPV, virus replication occurred in the lungs and spleens of infected hamsters, a neutralizing antibody was produced in some hamsters within 8 days post-challenge, and no conspicuous signs of disease occurred. CedPV replicated to a similar magnitude as NiV-Bangladesh in type I IFN-deficient BHK-21 Syrian hamster fibroblasts but replicated 4 logs lower in type I IFN-competent primary Syrian hamster and human pulmonary endothelial cells, a principal target of henipaviruses. The coinfection of these cells with CedPV and NiV failed to rescue CedPV titers and did not diminish NiV titers, suggesting the replication machinery is virus-specific. Type I IFN response transcripts Ifna7, Ddx58, Stat1, Stat2, Ccl5, Cxcl10, Isg20, Irf7, and Iigp1 were all significantly elevated in CedPV-infected hamster endothelial cells, whereas Ifna7 and Iigp1 expression were significantly repressed during NiV infection. These results are consistent with the hypothesis that CedPV's inability to counter the host type I IFN response may, in part, contribute to its lack of pathogenicity. Because NiV causes a fatal disease in Syrian hamsters with similarities to human disease, this model will provide valuable information about the pathogenic mechanisms of henipaviruses.


Subject(s)
Henipavirus Infections/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate , Virus Replication , Animals , Coinfection/immunology , Coinfection/virology , Cricetinae , Endothelial Cells/immunology , Endothelial Cells/virology , Female , Henipavirus/pathogenicity , Henipavirus/physiology , Humans , Lung/virology , Nipah Virus/pathogenicity , Nipah Virus/physiology , Spleen/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...